Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biomedicines ; 11(5)2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37238952

ABSTRACT

This study compares the effects of pre-processing multiple polymers together to form a single-phase polymer alloy prior to amorphous solid dispersion formulation. KinetiSol compounding was used to pre-process a 1:1 (w/w) ratio of hypromellose acetate succinate and povidone to form a single-phase polymer alloy with unique properties. Ivacaftor amorphous solid dispersions comprising either a polymer, an unprocessed polymer blend, or the polymer alloy were processed by KinetiSol and examined for amorphicity, dissolution performance, physical stability, and molecular interactions. A polymer alloy ivacaftor solid dispersion with a drug loading of 50% w/w was feasible versus 40% for the other compositions. Dissolution in fasted simulated intestinal fluid revealed that the 40% ivacaftor polymer alloy solid dispersion reached a concentration of 595 µg/mL after 6 h, 33% greater than the equivalent polymer blend dispersion. Fourier transform infrared spectroscopy and solid-state nuclear magnetic resonance revealed changes in the ability of the povidone contained in the polymer alloy to hydrogen bond with the ivacaftor phenolic moiety, explaining the differences in the dissolution performance. This work demonstrates that the creation of polymer alloys from polymer blends is a promising technique that provides the ability to tailor properties of a polymer alloy to maximize the drug loading, dissolution performance, and stability of an ASD.

2.
Eur J Pharm Biopharm ; 165: 52-65, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33979662

ABSTRACT

Abiraterone is a poorly water-soluble drug used in the treatment of prostate cancer. In our previous study, we reported that KinetiSol® processed solid dispersions (KSDs) based on hydroxypropyl ß-cyclodextrin (HPBCD) showed improved dissolution and pharmacokinetics of abiraterone. However, the nature of abiraterone-HPBCD interaction within the KSDs or the effect of drug loading on the physicochemical properties and in vivo performance of HPBCD-based KSDs remain largely unknown. We hypothesize that KinetiSol technology can prepare abiraterone-HPBCD complexes within KSDs and that increasing the drug loading beyond an optimal point reduces the in vitro and in vivo performance of these KSDs. To confirm our hypothesis, we developed KSDs with 10-50% w/w drug loading and analyzed them using X-ray diffractometry and modulated differential scanning calorimetry. We found that KSDs containing 10-30% drug were amorphous. Interestingly, two-dimensional solid-state nuclear magnetic resonance and Raman spectroscopy indicated that the abiraterone-HPBCD complexes were formed. At elevated temperatures, the 10% and 20% drug-loaded KSDs were physically stable, while the 30% drug-loaded KSD showed recrystallization of abiraterone. In vitro dissolution and in vivo pharmacokinetic performances improved as the drug loading decreased; we attribute this to increased noncovalent interactions between abiraterone and HPBCD at lower drug loadings. Overall, the 10% drug loaded KSD showed a dissolution enhancement of 15.7-fold compared to crystalline abiraterone, and bioavailability enhancement of 3.9-fold compared to the commercial abiraterone acetate tablet Zytiga®. This study is first to confirm that KinetiSol, a high-energy, solvent-free technology, is capable of forming abiraterone-HPBCD complexes. Furthermore, in terms of in vitro and in vivo performance, a 10% drug load is optimal.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/chemistry , Androstenes/pharmacokinetics , Drug Compounding/methods , Excipients/chemistry , Androstenes/chemistry , Biological Availability , Chemistry, Pharmaceutical , Drug Liberation
3.
Int J Pharm X ; 3: 100092, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34977559

ABSTRACT

We seek to further addresss the questions posed by Moseson et al. regarding whether any residual crystal level, size, or characteristic is acceptable in an amorphous solid dispersion (ASD) such that its stability, enhanced dissolution, and increased bioavailability are not compromised. To address this highly relevant question, we study an interesting heat- and shear-labile drug in development, LY3009120. To study the effects of residual crystallinity and degradation in ASDs, we prepared three compositionally identical formulations (57-1, 59-4, and 59-5) using the KinetiSol process under various processing conditions to obtain samples with various levels of crystallinity (2.3%, 0.9%, and 0.1%, respectively) and degradation products (0.74%, 1.97%, and 3.12%, respectively). Samples with less than 1% crystallinity were placed on stability, and we observed no measurable change in the drug's crystallinity, dissolution profile or purity in the 59-4 and 59-5 formulations over four months of storage under closed conditions at 25 °C and 60% humidity. For formulations 57-1, 59-4, and 59-5, bioavailability studies in rats reveal a 44-fold, 55-fold, and 62-fold increase in mean AUC, respectively, compared to the physical mixture. This suggests that the presence of some residual crystals after processing can be acceptable and will not change the properties of the ASD over time.

4.
AAPS PharmSciTech ; 21(8): 319, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33179174

ABSTRACT

We report for the first time that incorporation of a thermally conductive excipient (TCE) modifies the thermal conductivity of the ternary drug-polymer-TCE compositions such that high-energy mixing can occur for prolonged periods at a selected steady-state temperature during the KinetiSol process. In this study, candurin, a TCE, is incorporated within a composition that is processed by high-energy mixing from the KinetiSol process to increase the thermal conductivity of the ternary composition. The improved thermal conductivity promotes heat transfer and enables the high-energy mixing applied during the KinetiSol process to be continued for prolonged time intervals at a selected steady-state temperature, instead of undergoing a continued increase in temperature when the TCE is not present in the composition. The addition of candurin does not impact the molecular structure and mixing of the drug and polymer in ASDs from solid-state NMR characterizations. Compositions with candurin achieved a steady-state processing temperature with + 5°C of the target temperature, and these compositions demonstrated the ability to mix for prolonged time periods while maintaining within this steady-state temperature range, thus enabling the formation of an ASD at a temperature that the drug does not chemically degrade. This study demonstrated that inclusion of the TCE modified the composition's thermal conductivity to efficiently dissipate heat to achieve a selected steady-state temperature during the KinetiSol process, thus providing prolonged mixing times at a lower temperature for dissolving the drug into the polymer to achieve an ASD without sacrificing product performance.


Subject(s)
Drug Compounding/methods , Excipients/chemistry , Thermal Conductivity , Chemistry, Pharmaceutical/methods , Kinetics , Polymers/chemistry , Solubility
5.
AAPS PharmSciTech ; 21(8): 312, 2020 Nov 08.
Article in English | MEDLINE | ID: mdl-33161479

ABSTRACT

Thermal processing has gained much interest in the pharmaceutical industry, particularly for the enhancement of solubility, bioavailability, and dissolution of active pharmaceutical ingredients (APIs) with poor aqueous solubility. Formulation scientists have developed various techniques which may include physical and chemical modifications to achieve solubility enhancement. One of the most commonly used methods for solubility enhancement is through the use of amorphous solid dispersions (ASDs). Examples of commercialized ASDs include Kaletra®, Kalydeco®, and Onmel®. Various technologies produce ASDs; some of the approaches, such as spray-drying, solvent evaporation, and lyophilization, involve the use of solvents, whereas thermal approaches often do not require solvents. Processes that do not require solvents are usually preferred, as some solvents may induce toxicity due to residual solvents and are often considered to be damaging to the environment. The purpose of this review is to provide an update on recent innovations reported for using hot-melt extrusion and KinetiSol® Dispersing technologies to formulate poorly water-soluble APIs in amorphous solid dispersions. We will address development challenges for poorly water-soluble APIs and how these two processes meet these challenges.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Compounding/methods , Hot Temperature , Biological Availability , Drug Carriers , Excipients , Solubility , Water
6.
Mol Pharm ; 17(8): 2789-2808, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32520562

ABSTRACT

Oral delivery of poorly water-soluble, weakly basic drugs may be problematic based on the drugs' intrinsic properties. Many drugs in this subset have overcome barriers to delivery following successful formulation as amorphous solid dispersions (ASDs). To process drugs as ASDs, multiple commercially relevant technologies have been developed and become well understood. However, ASD-producing technologies like spray drying and KinetiSol produce ASDs with vastly differing particle characteristics. Ultimately, the objective of this study was to assess whether processing an ASD of identical composition utilizing two different ASD-producing technologies (KinetiSol and spray drying) may impact the oral bioavailability of a weakly basic drug. For this study, we selected a weakly basic drug (Boehringer Ingelheim research compound 639667, BI 667) and processed it with an anionic polymer (hypromellose acetate succinate MMP grade (HPMCAS-MMP)) to evaluate whether the processing technology could modulate drug release in acidic and neutral media. Multiple characterization techniques (specific surface area (SSA), particle size distribution (PSD), scanning electron microscopy (SEM)) were utilized to evaluate the surface characteristics and differences in particles produced by KinetiSol and spray drying. Molecular interactions and drug-polymer miscibility of the processed particles were assessed using Fourier transform infrared spectroscopy and solid-state nuclear magnetic resonance, respectively. In vitro nonsink, pH-shift dissolution in biorelevant media and dissolution/permeation studies were conducted to better understand the release of BI 667 based on processing technology and particle size distribution. Finally, an in vivo male Beagle dog study was conducted to assess the impact of processing technology on oral bioavailability. In this study, we demonstrate that particles produced by KinetiSol have enhanced oral bioavailability compared with spray-dried particles when delivering a weakly basic drug processed with an anionic polymer. The findings of this study demonstrate that by utilizing KinetiSol, drug release may be controlled such that supersaturation in acidic media is inhibited and supersaturation of the drug is designed to occur in neutral media, ultimately enhancing oral bioavailability.


Subject(s)
Pharmaceutical Preparations/chemistry , Polymers/chemistry , Animals , Chemistry, Pharmaceutical/methods , Dogs , Drug Carriers/chemistry , Drug Compounding/methods , Drug Liberation/drug effects , Male , Solubility/drug effects , Water/chemistry
7.
Biochim Biophys Acta Rev Cancer ; 1873(1): 188319, 2020 01.
Article in English | MEDLINE | ID: mdl-31678141

ABSTRACT

The treatment of cancer is still a major challenge. But tremendous progress in anticancer drug discovery and development has occurred in the last few decades. However, this progress has resulted in few effective oncology products due to challenges associated with anticancer drug delivery. Oral administration is the most preferred route for anticancer drug delivery, but the majority of anticancer drugs currently in product pipelines and the majority of those that have been commercially approved have inherently poor water solubility, and this cannot be mitigated without compromising their potency and stability. The poor water solubility of anticancer drugs, in conjunction with other factors, leads to suboptimal pharmacokinetic performance. Thus, these drugs have limited efficacy and safety when administered orally. The amorphous solid dispersion (ASD) is a promising formulation technology that primarily enhances the aqueous solubility of poorly water-soluble drugs. In this review, we discuss the challenges associated with the oral administration of anticancer drugs and the use of ASD technology in alleviating these challenges. We emphasize the ability of ASDs to improve not only the pharmacokinetics of poorly water-soluble anticancer drugs, but also their efficacy and safety. The goal of this paper is to rationalize the application of ASD technology in the formulation of anticancer drugs, thereby creating superior oncology products that lead to improved therapeutic outcomes.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Drug Monitoring/methods , Technology, Pharmaceutical/methods , Administration, Oral , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Chemistry, Pharmaceutical , Drug Delivery Systems/trends , Drug Discovery/methods , Drug Discovery/trends , Drug Monitoring/trends , Humans , Solubility , Technology, Pharmaceutical/trends , Water/chemistry
8.
AAPS PharmSciTech ; 19(5): 1985-1997, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29869311

ABSTRACT

Ritonavir is an anti-viral compound that has also been employed extensively as a CYP3A4 and P-glycoprotein (Pgp) inhibitor to boost the pharmacokinetic performance of compounds that undergo first pass metabolism. For use in combination products, there is a desire to minimize the mass contribution of the ritonavir system to reduce patient pill burden in these combination products. In this study, KinetiSol® processing was utilized to produce an amorphous solid dispersion of ritonavir at two times the drug load of the commercially available form of ritonavir, and the composition was subsequently developed into a tablet dosage form. The amorphous intermediate was demonstrated to be amorphous by X-ray powder diffraction and 13C solid-state nuclear magnetic resonance and an intimately mixed single-phase system by modulated differential scanning calorimetry and 1H T1/1H T1ρ solid-state nuclear magnetic resonance relaxation. In vitro transmembrane flux analysis showed similar permeation rates for the KinetiSol-made tablet and the reference tablet dosage form, Norvir®. In vivo pharmacokinetic comparison between the two dosage forms resulted in equivalent exposure with approximately 20% Cmax reduction for the KinetiSol tablet. These performance gains were realized with a concurrent reduction in dosage form mass of 45%.


Subject(s)
HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/chemistry , Ritonavir/administration & dosage , Ritonavir/chemistry , Acids/chemistry , Animals , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical/methods , Dogs , Drug Compounding/methods , HIV Protease Inhibitors/pharmacokinetics , Magnetic Resonance Spectroscopy , Permeability , Ritonavir/pharmacokinetics , Tablets , X-Ray Diffraction
10.
AAPS PharmSciTech ; 19(5): 1933-1956, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29846889

ABSTRACT

Due to the high number of poorly soluble drugs in the development pipeline, novel processes for delivery of these challenging molecules are increasingly in demand. One such emerging method is KinetiSol, which utilizes high shear to produce amorphous solid dispersions. The process has been shown to be amenable to difficult to process active pharmaceutical ingredients with high melting points, poor organic solubility, or sensitivity to heat degradation. Additionally, the process enables classes of polymers not conventionally processable due to their high molecular weight and/or poor organic solubility. Beyond these advantages, the KinetiSol process shows promise with other applications, such as the production of amorphous mucoadhesive dispersions for delivery of compounds that would also benefit from permeability enhancement.


Subject(s)
Chemistry, Pharmaceutical/methods , Dosage Forms , Drug Compounding/methods , Animals , Biological Availability , Desiccation , Drug Carriers , Humans , Pharmaceutical Preparations/chemistry
11.
AAPS PharmSciTech ; 19(5): 1957-1970, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29541940

ABSTRACT

Vemurafenib is a poorly soluble, low permeability drug that has a demonstrated need for a solubility-enhanced formulation. However, conventional approaches for amorphous solid dispersion production are challenging due to the physiochemical properties of the compound. A suitable and novel method for creating an amorphous solid dispersion, known as solvent-controlled coprecipitation, was developed to make a material known as microprecipitated bulk powder (MBP). However, this approach has limitations in its processing and formulation space. In this study, it was hypothesized that vemurafenib can be processed by KinetiSol into the same amorphous formulation as MBP. The KinetiSol process utilizes high shear to rapidly process amorphous solid dispersions containing vemurafenib. Analysis of the material demonstrated that KinetiSol produced amorphous, single-phase material with acceptable chemical purity and stability. Values obtained were congruent to analysis conducted on the comparator material. However, the materials differed in particle morphology as the KinetiSol material was dense, smooth, and uniform while the MBP comparator was porous in structure and exhibited high surface area. The particles produced by KinetiSol had improved in-vitro dissolution and pharmacokinetic performance for vemurafenib compared to MBP due to slower drug nucleation and recrystallization which resulted in superior supersaturation maintenance during drug release. In the in-vivo rat pharmacokinetic study, both amorphous solid dispersions produced by KinetiSol exhibited mean AUC values at least two-fold that of MBP when dosed as a suspension. It was concluded that the KinetiSol process produced superior dosage forms containing vemurafenib with the potential for substantial reduction in patient pill burden.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Drug Compounding/methods , Indoles/chemistry , Indoles/pharmacokinetics , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Animals , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical/methods , Crystallization , Desiccation , Dosage Forms , Drug Stability , Male , Particle Size , Powders , Rats , Rats, Sprague-Dawley , Solubility , Vemurafenib , X-Ray Diffraction
12.
AAPS PharmSciTech ; 19(5): 1998-2008, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29192405

ABSTRACT

The formulation factors relevant to developing immediate and controlled release dosage forms containing poorly soluble drugs dispersed in amorphous systems are poorly understood. While the utility of amorphous solid dispersions is becoming apparent in the pharmaceutical marketplace, literature reports tend to concentrate on the development of solid dispersion particulates, which then must be formulated into a tablet. Amorphous solid dispersions of itraconazole in high molecular weight hydroxypropyl methylcellulose were prepared by KinetiSol® Dispersing and tablets were formulated to immediately disintegrate or control the release of itraconazole. Formulated tablets were evaluated by two non-sink dissolution methodologies and the dosage form properties that controlled the gelling tendency of the dispersion carrier, hydroxypropyl methylcellulose, were investigated. Selected formulations were evaluated in an exploratory beagle dog pharmacokinetic study; the results of which indicate potential for a prolonged absorption phase relative to the commercially extruded control.


Subject(s)
Antifungal Agents/administration & dosage , Antifungal Agents/chemistry , Itraconazole/administration & dosage , Itraconazole/chemistry , Animals , Antifungal Agents/pharmacokinetics , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical/methods , Dogs , Drug Compounding/methods , Excipients , Hydrogen-Ion Concentration , Hypromellose Derivatives , Intestinal Mucosa/metabolism , Itraconazole/pharmacokinetics , Molecular Weight , Solubility , Tablets , X-Ray Diffraction
13.
Eur J Pharm Biopharm ; 113: 157-167, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28088635

ABSTRACT

The oral delivery of mucoadhesive patches has been shown to enhance the absorption of large molecules such as peptides. We hypothesized that this mechanism could have utility for poorly soluble small molecules by utilizing a mucoadhesive polymer as the matrix for an amorphous solid dispersion. Binary dispersions of itraconazole and carbomer (Carbopol 71G) were prepared utilizing a thermokinetic mixing process (KinetiSol Dispersing) and the physicochemical properties were investigated by powder X-ray diffraction, calorimetry, and liquid chromatography. Adhesion of the dispersions to freshly excised porcine intestine was investigated with a texture analyzer. Minitablets were compressed from the optimal dispersion and further investigated in vitro and in vivo in rats. Thermokinetic mixing successfully processed amorphous dispersions up to 30% drug loading and each dispersion exhibited works of adhesion that were approximately an order of magnitude greater than a negative control in vitro. Ethylcellulose (EC) coated and uncoated minitablets prepared with the 30% drug load dispersion were delivered orally to rats and exhibited sustained release characteristics, with overall bioavailability greater for the uncoated minitablets compared to the EC-coated minitablets, similar to the rank order observed in our in vitro dissolution experiments. Necropsy studies showed that minitablets delivered with enteric-coated capsules targeted release to the distal small intestine and adhered to the intestinal mucosa, but the rat model presented limitations with respect to evaluating the overall performance. Based on the in vitro and in vivo results, further investigations in larger animals are a logical next step where fluid volumes, pH, and transit times are more favorable for the evaluated dosage forms.


Subject(s)
Antifungal Agents/administration & dosage , Itraconazole/administration & dosage , Animals , Antifungal Agents/chemistry , Antifungal Agents/pharmacokinetics , Calorimetry , Chromatography, Liquid , Itraconazole/chemistry , Itraconazole/pharmacokinetics , Powder Diffraction , Rats , Rats, Sprague-Dawley , Solubility , Tablets
14.
Eur J Pharm Biopharm ; 101: 72-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26861929

ABSTRACT

Polyvinyl alcohol has received little attention as a matrix polymer in amorphous solid dispersions (ASDs) due to its thermal and rheological limitations in extrusion processing and limited organic solubility in spray drying applications. Additionally, in extrusion processing, the high temperatures required to process often exclude thermally labile APIs. The purpose of this study was to evaluate the feasibility of processing polyvinyl alcohol amorphous solid dispersions utilizing the model compound ritonavir with KinetiSol® Dispersing (KSD) technology. The effects of KSD rotor speed and ejection temperature on the physicochemical properties of the processed material were evaluated. Powder X-ray diffraction and modulated differential scanning calorimetry were used to confirm amorphous conversion. Liquid chromatography-mass spectroscopy was used to characterize and identify degradation pathways of ritonavir during KSD processing and (13)C nuclear magnetic resonance spectroscopy was used to investigate polymer stability. An optimal range of processing conditions was found that resulted in amorphous product and minimal to no drug and polymer degradation. Drug release of the ASD produced from the optimal processing conditions was evaluated using a non-sink, pH-shift dissolution test. The ability to process amorphous solid dispersions with polyvinyl alcohol as a matrix polymer will enable further investigations of the polymer's performance in amorphous systems for poorly water-soluble compounds.


Subject(s)
Drug Compounding/methods , Polyvinyl Alcohol/chemistry , Ritonavir/chemistry , Calorimetry, Differential Scanning/methods , Chemistry, Pharmaceutical/methods , Desiccation , Drug Carriers/chemistry , Drug Stability , Excipients/chemistry , Hot Temperature , Polymers/chemistry , Solubility , Temperature , Water/chemistry , X-Ray Diffraction/methods
15.
AAPS PharmSciTech ; 17(1): 180-90, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26863889

ABSTRACT

The KinetiSol® Dispersing (KSD) technology has enabled the investigation into the use of polyvinyl alcohol (PVAL) as a concentration enhancing polymer for amorphous solid dispersions. Our previous study revealed that the 88% hydrolyzed grade of PVAL was optimal for itraconazole (ITZ) amorphous compositions with regard to solid-state properties, non-sink dissolution performance, and bioavailability enhancement. The current study investigates the influence of molecular weight for the 88% hydrolyzed grades of PVAL on the properties of KSD processed ITZ:PVAL amorphous dispersions. Specifically, molecular weights in the processable range of 4 to 18 mPa · s were evaluated and the 4-88 grade provided the highest AUC dissolution profile. Amorphous dispersions at 10, 20, 30, 40, and 50% ITZ drug loads in PVAL 4-88 were also compared by dissolution performance. Analytical tools of diffusion-ordered spectroscopy and Fourier transform infrared spectroscopy were employed to understand the interaction between drug and polymer. Finally, results from a 30-month stability test of a 30% drug loaded ITZ:PVAL 4-88 composition shows that stable amorphous dispersions can be achieved. Thus, this newly enabled polymer carrier can be considered a viable option for pharmaceutical formulation development for solubility enhancement.


Subject(s)
Itraconazole/chemistry , Polymers/chemistry , Polyvinyl Alcohol/chemistry , Solubility , Water/chemistry , Biological Availability , Chemistry, Pharmaceutical/methods , Drug Compounding/methods , Drug Delivery Systems/methods , Excipients/chemistry , Itraconazole/pharmacokinetics
16.
AAPS PharmSciTech ; 17(1): 120-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26729526

ABSTRACT

Thermal processing technologies continue to gain interest in pharmaceutical manufacturing. However, the types and grades of polymers that can be utilized in common thermal processing technologies, such as hot-melt extrusion (HME), are often limited by thermal or rheological factors. The objectives of the present study were to compare and contrast two thermal processing methods, HME and KinetiSol® Dispersing (KSD), and investigate the influence of polymer type, polymer molecular weight, and drug loading on the ability to produce amorphous solid dispersions (ASDs) containing the model compound griseofulvin (GRIS). Dispersions were analyzed by a variety of imaging, solid-state, thermal, and solution-state techniques. Dispersions were prepared by both HME and KSD using polyvinylpyrrolidone (PVP) K17 or hydroxypropyl methylcellulose (HPMC) E5. Dispersions were only prepared by KSD using higher molecular weight grades of HPMC and PVP, as these could not be extruded under the conditions selected. Powder X-ray diffraction (PXRD) analysis showed that dispersions prepared by HME were amorphous at 10% and 20% drug load; however, it showed significant crystallinity at 40% drug load. PXRD analysis of KSD samples showed all formulations and drug loads to be amorphous with the exception of trace crystallinity seen in PVP K17 and PVP K30 samples at 40% drug load. These results were further supported by other analytical techniques. KSD produced amorphous dispersions at higher drug loads than could be prepared by HME, as well as with higher molecular weight polymers that were not processable by HME, due to its higher rate of shear and torque output.


Subject(s)
Drug Compounding/methods , Lactose/analogs & derivatives , Methylcellulose/analogs & derivatives , Polymers/chemistry , Polyvinyls/chemistry , Pyrrolidines/chemistry , Chemistry, Pharmaceutical/methods , Drug Carriers/chemistry , Excipients/chemistry , Griseofulvin/chemistry , Hot Temperature , Lactose/chemistry , Methylcellulose/chemistry , Molecular Weight , Solutions/chemistry , X-Ray Diffraction/methods
17.
AAPS PharmSciTech ; 17(1): 167-79, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26637232

ABSTRACT

Polyvinyl alcohol (PVAL) has not been investigated in a binary formulation as a concentration-enhancing polymer owing to its high melting point/high viscosity and poor organic solubility. Due to the unique attributes of the KinetiSol® dispersing (KSD) technology, PVAL has been enabled for this application and it is the aim of this paper to investigate various grades for improvement of the solubility and bioavailability of poorly water soluble active pharmaceutical ingredients. Solid amorphous dispersions were created with the model drug, itraconazole (ITZ), at a selected drug loading of 20%. Polymer grades were chosen with variation in molecular weight and degree of hydroxylation to determine the effects on performance. Differential scanning calorimetry, powder X-ray diffraction, polarized light microscopy, size exclusion chromatography, and dissolution testing were used to characterize the amorphous dispersions. An in vivo pharmacokinetic study in rats was also conducted to compare the selected formulation to current market formulations of ITZ. The 4-88 grade of PVAL was determined to be effective at enhancing solubility and bioavailability of itraconazole.


Subject(s)
Itraconazole/chemistry , Polymers/chemistry , Polyvinyl Alcohol/chemistry , Solubility , Water/chemistry , Animals , Biological Availability , Calorimetry, Differential Scanning/methods , Chemistry, Pharmaceutical/methods , Drug Compounding/methods , Drug Delivery Systems/methods , Excipients/chemistry , Itraconazole/pharmacokinetics , Male , Powders/chemistry , Rats , Rats, Sprague-Dawley , X-Ray Diffraction/methods
18.
J Pharm Pharmacol ; 68(5): 678-91, 2016 May.
Article in English | MEDLINE | ID: mdl-26454138

ABSTRACT

OBJECTIVES: To evaluate the effect of ritonavir (RTV) co-administration on the bioavailability of an amorphous dispersion of acetyl-11-keto-beta-boswellic acid (AKBA) and to develop a pharmaceutically acceptable AKBA-RTV combination tablet. METHODS: A pharmacokinetic (PK) study in rats was conducted to evaluate the influence of RTV co-administration on the oral bioavailability of an AKBA amorphous dispersion. KinetiSol was utilized to enable production of an improved RTV formulation that facilitated the development of an AKBA-RTV combination tablet. Following in-vitro characterization, the PK performance of the tablets was evaluated in male beagles. KEY FINDINGS: Co-administration of RTV increased oral absorption of AKBA by about fourfold over the AKBA dispersion alone and approximately 24-fold over the pure compound. The improved RTV amorphous dispersion exhibited similar purity and neutral-phase dissolution to Norvir. The AKBA-RTV combination tablets yielded a substantial increase in AKBA's bioavailability in dogs. CONCLUSIONS: Oral absorption of AKBA is substantially limited by intestinal CYP3A activity and poor aqueous solubility. Consequently, AKBA's oral bioavailability is maximized by administration from a supersaturating formulation in conjunction with a CYP3A inhibitor. The AKBA-RTV combination tablet presented herein represents a breakthrough in the oral delivery of the compound facilitating future use as a drug therapy for broad spectrum cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Intestines/drug effects , Ritonavir/pharmacokinetics , Triterpenes/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Biological Availability , Biotransformation , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/chemistry , Dogs , Drug Combinations , Drug Compounding , Intestines/enzymology , Male , Ritonavir/administration & dosage , Ritonavir/chemistry , Tablets , Technology, Pharmaceutical/methods , Triterpenes/administration & dosage , Triterpenes/chemistry
19.
Drug Dev Ind Pharm ; 41(3): 382-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24329130

ABSTRACT

Acetyl-11-keto-ß-boswellic acid (AKBA), a gum resin extract, possesses poor water-solubility that limits bioavailability and a high melting point making it difficult to successfully process into solid dispersions by fusion methods. The purpose of this study was to investigate solvent and thermal processing techniques for the preparation of amorphous solid dispersions (ASDs) exhibiting enhanced solubility, dissolution rates and bioavailability. Solid dispersions were successfully produced by rotary evaporation (RE) and KinetiSol® Dispersing (KSD). Solid state and chemical characterization revealed that ASD with good potency and purity were produced by both RE and KSD. Results of the RE studies demonstrated that AQOAT®-LF, AQOAT®-MF, Eudragit® L100-55 and Soluplus with the incorporation of dioctyl sulfosuccinate sodium provided substantial solubility enhancement. Non-sink dissolution analysis showed enhanced dissolution properties for KSD-processed solid dispersions in comparison to RE-processed solid dispersions. Variances in release performance were identified when different particle size fractions of KSD samples were analyzed. Selected RE samples varying in particle surface morphologies were placed under storage and exhibited crystalline growth following solid-state stability analysis at 12 months in comparison to stored KSD samples confirming amorphous instability for RE products. In vivo analysis of KSD-processed solid dispersions revealed significantly enhanced AKBA absorption in comparison to the neat, active substance.


Subject(s)
Chemistry, Pharmaceutical/methods , Frankincense/chemical synthesis , Plant Gums/chemical synthesis , Triterpenes/chemical synthesis , Water/chemistry , Animals , Frankincense/metabolism , Male , Plant Gums/metabolism , Rats , Rats, Sprague-Dawley , Solubility , Triterpenes/metabolism , Water/metabolism , X-Ray Diffraction
20.
Eur J Pharm Sci ; 48(4-5): 758-66, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23348153

ABSTRACT

The dissolution enhancement advantages inherent to amorphous solid dispersions systems are often not fully realized once they are formulated into a solid dosage form. The objective of this study was to investigate the ability of inorganic salts to improve the dissolution rate of carbamazepine (CBZ) from tablets containing a high loading of a Soluplus®-based solid dispersion. Cloud point and viscometric studies were conducted on Soluplus® solutions to understand the effect of temperature, salt type and salt concentration on the aqueous solubility and gelling tendencies of Soluplus®, properties that can significantly impact dissolution performance. Studies indicated that Soluplus® exhibited a cloud point that was strongly dependent on the salt type and salt concentration present in the dissolving medium. The presence of kosmotropic salts dehydrated the polymer, effectively lowering the cloud point and facilitating formation of a thermoreversible hydrogel. The ability of ions to impact the cloud point and gel strength generally followed the rank order of the Hofmeister series. Solid dispersions of CBZ and Soluplus® were prepared by KinetiSol® Dispersing, characterized to confirm an amorphous composition was formed and incorporated into tablets at very high levels (70% w/w). Dissolution studies demonstrated the utility of including salts in tablets to improve dissolution properties. Tablets that did not contain a salt or those that included a chaotropic salt hydrated at the tablet surface and did not allow for sufficient moisture ingress into the tablet. Conversely, the inclusion of kosmotropic salts allowed for rapid hydration of the entire tablet and the formation of a gel structure with strength dependent on the type of salt utilized. Studies also showed that, in addition to allowing tablet hydration, potassium bicarbonate and potassium carbonate provided effervescence which effectively destroyed the gel network and allowed for rapid dissolution of CBZ. Subsequent dissolution studies in 0.1 N HCl showed that potassium bicarbonate was an effective tablet disintegrant at levels as low as 1% and provided for tablets that rapidly disintegrated over a wide range of applied compression forces, presumably due to synergy between the ability to form a weak hydrogel structure and carbon dioxide liberation. Similar dissolution performance was measured in pH 4.5 acetate buffer, despite reduced polymer solubility caused by kosmotropic salts in solution, demonstrating robustness. With the use of inorganic salts such as potassium bicarbonate, it may be possible to substantially improve disintegration and dissolution characteristics of tablets containing Soluplus®.


Subject(s)
Carbamazepine/chemistry , Excipients/chemistry , Polyethylene Glycols/chemistry , Polyvinyls/chemistry , Salts/chemistry , Powder Diffraction , Solubility , Tablets , Viscosity , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...